Abstract
Patients with cystic fibrosis (CF) commonly have lower circulating concentrations of 25-hydroxyvitamin D (25(OH)D) than healthy populations. We comprehensively compared measures of vitamin D metabolism among individuals with CF and healthy control subjects. In a cross-sectional study, serum from participants with CF (N=83) and frequency-matched healthy control subjects by age and race (N=82) were analyzed for: 25(OH)D2 and 25(OH)D3, 1α,25-dihydroxyvitamins D2 and D3 (1α,25(OH)2D2 and 1α,25(OH)2D3), 24,25-dihydroxyvitamin D3 (24,25(OH)2D3), 4β,25-dihydroxyvitamin D3 (4β,25(OH)2D3), 25-hydroxyvitamin D3-3-sulfate (25(OH)D3-S), and 25-hydroxyvitamin D3-3-glucuronide (25(OH)D3-G). In a 56-day prospective pharmacokinetic study, ~25 μg deuterium-labeled 25(OH)D3 (d6-25(OH)D3) was administered intravenously to participants (N=5 with CF, N=5 control subjects). Serum was analyzed for d6-25(OH)D3 and d6-24,25(OH)2D3, and pharmacokinetic parameters were estimated. In the cross-sectional study, participants with CF had similar mean (SD) total 25(OH)D concentrations as control subjects (26.7 [12.3] vs. 27.7 [9.9] ng/mL) and had higher vitamin D supplement use (53% vs. 22%). However, participants with CF had lower total 1α,25(OH)2D (43.6 [12.7] vs. 50.7 [13.0] pg/mL), 4β,25(OH)2D3 (52.1 [38.9] vs. 79.9 [60.2] pg/mL), and 25(OH)D3-S (17.7 [11.6] vs. 30.1 [12.3] ng/mL) (p < 0.001 for all). The pharmacokinetics of d6-25(OH)D3 and d6-24,25(OH)D3 did not differ between groups. In summary, although 25(OH)D concentrations were comparable, participants with CF had lower 1α,25(OH)2D, 4β,25(OH)2D3, and 25(OH)D3-S concentrations than healthy controls. Neither 25(OH)D3 clearance, nor formation of 24,25(OH)2D3, appears to account for these differences and alternative mechanisms for low 25(OH)D in CF (i.e., decreased formation, altered enterohepatic recirculation) should be explored.
Keywords: Cystic fibrosis; 25-hydroxyvitamin D; 24, 25-dihydroxyvitamin D; 1α,25dihydroxyvitamin D; pharmacokinetics
1. Introduction
Vitamin D deficiency, defined by low circulating concentrations of 25-hydroxyvitamin D (25(OH)D), is ubiquitous in patients with cystic fibrosis (CF) (1). Most patients with this disorder require vitamin D supplementation, often at high doses, to maintain physiologically normal 25(OH)D concentrations. Vitamin D deficiency in patients with CF is associated with low bone mineral density, impaired innate and acquired immunity, and increased risk of pulmonary complications (1–3). These problems are further compounded due to decreased ability to exercise, malnutrition due to poor absorption of nutrients, and use of corticosteroids (2, 3). The Cystic Fibrosis Foundation recommends a target 25(OH)D of ≥ 30 ng/mL (75 nM) (4). However, up to 90% of persons with CF do not achieve this recommended threshold despite routine supplementation (2, 4–7), and the specific mechanisms by which their 25(OH)D concentrations remain low are poorly understood.
Vitamin D exists in two primary forms: vitamin D2 and vitamin D3, both of which follow similar pathways of metabolism (6, 8) (Figure S1). Derived mostly from sunlight, vitamin D is converted in the liver to 25(OH)D, the most widely used biomarker of long-term vitamin D exposure due to its long plasma half-life of 2 to 3 weeks (8, 9). 25(OH)D3, which is more abundant than 25(OH)D2, undergoes metabolism by cytochrome P450 (CYP) 27B1, also called 1α-hydroxylase, to form the biologically active 1α,25-dihydroxyvitamin D3 (1α,25(OH)2D3) (7, 10) (Figure S1). 25(OH)D3 may also be cleared from the blood through hydroxylation or conjugation into several inactive metabolites (11): 24R,25-dihydroxyvitamin D3 (24,25(OH)2D3, the most abundant metabolite of 25(OH)D clearance) (7, 10), 1β,25-dihydroxyvitamin D3 (12), 4β,25-dihydroxyvitamin D3 (4β,25(OH)2D3) (13), 25-hydroxyvitamin D3-3-sulfate (25(OH)D3-S) (14), and 25-hydroxyvitamin D3-3-glucuronide (25(OH)D3-G) (15) (Figure S1).
Numerous mechanisms for low 25(OH)D in persons with CF have been hypothesized: reduced time outdoors, poor intestinal absorption of fat-soluble vitamins such as vitamin D, reduced capacity to hydroxylate vitamin D in the liver, reduced storage capacity due to low body fat (1), impaired enterohepatic recirculation of vitamin D metabolites enhancing their loss (18), urinary loss of vitamin D binding protein (VDBP) and albumin (the major carriers of vitamin D metabolites) (16), and increased 1α-hydroxylase expression in pulmonary epithelial cells that drive 1α,25(OH)2D formation from 25(OH)D (17). Understanding the specific pathways that contribute to low circulating 25(OH)D concentrations in CF may improve how we assess and treat vitamin D-related complications of CF. Thus, we investigated the hypothesis that individuals with CF have altered concentrations of multiple vitamin D metabolites and increased clearance of 25(OH)D.
2. Methods
2.1. Study Design
We conducted two complementary studies to increase the understanding of vitamin D metabolism in CF. First, we compared a comprehensive set of vitamin D metabolites between participants with CF and healthy control subjects in a cross-sectional study. Second, we determined the pharmacokinetics of vitamin D metabolism by administering intravenous deuterium-labeled 25-hydroxyvitamin D3 (d6-25(OH)D3) to participants with CF and healthy controls. Both studies were approved by the University of Washington Institutional Review Board. The pharmacokinetic study was approved by the US Food and Drug Administration’s Investigational New Drug program and registered on clinicaltrials.gov (NCT03104855).
2.1. Study Populations
For the cross-sectional study, we recruited participants ≥ 18 years of age with a clinical diagnosis of CF from specialty clinics at the University of Washington. All genotypes of CF were included. Participants were required to be clinically stable, defined as no hospitalization or intravenous antibiotics administered within two weeks of study visit. Healthy control subjects were identified from the Healthy Kidney Study, a cross-sectional study that previously collected biosamples and data using a similar protocol to serve as control subjects for a broad range of research studies (18, 19). Control subjects were frequency matched to CF participants by age and race.
We then invited participants with CF from the cross-sectional study to enroll in the prospective pharmacokinetic study. Healthy control subjects were recruited from the Seattle, WA area, frequency matched by age and race. For all participants in the pharmacokinetic study, inclusion criteria were age 18 years or older and a serum 25(OH)D concentration between 10–50 ng/mL. Participants were excluded if they had taken supplements of more than 400 IU/day of vitamin D3, any vitamin D2 supplements, 1α,25(OH)2D3 or analogues, calcimimetics within 3 months of enrollment, used vitamin D receptor agonists or cinacalcet within 4 weeks of enrollment, or used cytochrome P450 inhibitors or inducers within 4 weeks of enrollment. Participants on excluded drugs were allowed to washout before enrolling. Participants were also excluded for primary hyperparathyroidism, gastric bypass, tuberculosis, sarcoidosis, current pregnancy, hemoglobin < 10 g/dL, kidney transplant, or inability to provide informed consent.
2.2. Pharmacokinetic Study Design
We administered a single intravenous dose of d6-25(OH)D3 (26,26,26,27,27,27-d6; ISOTEC, Stable Isotope Division, Sigma-Aldrich, Miamisburg, OH) to participants with CF and healthy controls (9). The dose of d6-25(OH)D3 was individualized for each study participant and calculated to target a maximum initial concentration of d6-25(OH)D3 of 5 ng/mL. The dose was calculated as (5 ng/mL) × (estimated blood volume), where the estimated blood volume was calculated as described by Nadler et al. (20). Blood samples were collected at 15 minutes, 4 hours, 1, 4, 7, 14, 21, 28, 42, and 56 days after d6-25(OH)D3 administration. Serum was harvested by centrifugation and samples were stored at −80°C until analysis.
2.3. Vitamin D Metabolite Sample Analysis
We used dedicated liquid chromatography tandem mass spectrometry assays with internal standards and external calibration to quantify 25(OH)D2, 25(OH)D3, 1α,25(OH)2D2, 1α,25(OH)2D3, 24,25(OH)2D3, 4β,25(OH)2D3, 25(OH)D3-S, and 25(OH)D3-G (supplemental material). Total 25(OH)D and 1α,25(OH)2D were calculated by summing the concentrations of 25(OH)D2 and 25(OH)D3, or 1α,25(OH)2D2 and 1α,25(OH)2D3, respectively.
Free (i.e., unbound) concentrations of 25(OH)D3 and 1α,25(OH)2D3 were calculated using the previously published equation (21, 22),
where Dfree is the calculated free concentration of 25(OH)D3 or 1α,25(OH)2D3. Dtotal is the measured serum concentration of 25(OH)D3 or 1α,25(OH)2D3. [ALB] and [VDBP] are the measured serum albumin and VDBP concentrations, respectively. kALB and kVDPB are the association constants for albumin and VDBP, respectively (21–23). The values of the 25(OH)D3 association constants for albumin and VDBP are 6 × 105 M−1 and 7 × 108 M−1, respectively. The values of the 1α,25(OH)2D3 association constants for albumin and VDBP are 5.4 × 104 M−1 and 3.7 × 107 M−1, respectively. The percent unbound of 25(OH)D3 and 1α,25(OH)2D3 was estimated using % unbound = Dfree/Dtotal ·100%.
For the pharmacokinetic study, the liquid chromatography-mass spectrometry assays were adapted to quantify d6-25(OH)D3 and deuterium-labeled 24,25-dihydroxyvitamin D3 (d6-24,25(OH)2D3) in the serum samples (supplemental materials). In preliminary experiments, no other deuterated metabolites following d6-25(OH)D3 administration were detected (data not shown).
2.4. Pharmacokinetic Data Analysis
Serum concentrations of d6-25(OH)D3 and d6-24,25(OH)2D3 were analyzed using non-compartmental analysis with Phoenix WinNonlin software (version 8.0.0). Due to variability in the dose administered, the concentration-time profiles were dose-normalized and scaled to a 25 μg dose of d6-25(OH)D3. For d6-25(OH)D3, Cmax and Tmax were the observed dose-normalized peak d6-25(OH)D3 concentration and the time when the peak concentration was observed, respectively. The elimination rate constant (k) was estimated using uniform weighting of timepoints after 14 days. Elimination half-life of d6-25(OH)D3 was calculated as t1/2 = ln(2)/k. Area under the concentration-time curve (AUC0−∞) was calculated using the trapezoidal rule to calculate AUC0-last (to the last observed timepoint) and extrapolated to time = infinity. The percent of AUC extrapolated was calculated as AUC0-last/AUC0−∞ × 100. The systemic clearance (CL) of d6-25(OH)D3 was calculated as dose/AUC0−∞. The steady-state volume of distribution (Vss) was calculated as (dose∙AUMC)/(AUC)2, where the AUMC was calculated as the area under the first moments curve.
Concentrations of d6-24,25(OH)2D3 were not dose-normalized for pharmacokinetic analyses. The elimination rate constant of d6-24,25(OH)2D3 was not estimated as measured concentrations were near the lower limit of quantification resulting in too few timepoints describing decreasing d6-24,25(OH)2D3 concentrations. The molar AUC0-last ratio was calculated as (d6-24,25(OH)2D3 AUC0-last)/(d6-25(OH)D3 AUC0-last) using the observed d6-25(OH)D3 AUC0-last (not dose-normalized).
2.5. Statistical Analysis
We estimated the difference in concentrations of each vitamin D metabolite and other biomarkers between participants with CF and healthy controls using linear regression. Adjusted models included the variables of age, sex, and season or age, sex, season, and upstream vitamin D metabolite: total 25(OH)D for 1α,25(OH)2D, 25(OH)D3 for 25(OH)D3-derived metabolites, or 25(OH)D2 for 1α,25(OH)D2.
Pharmacokinetic parameters of participants with CF were compared to healthy controls using a Mann-Whitney U test. A value of p < 0.05 was considered statistically significant.
3. Results
3.1. Participant Characteristics
In total, 83 participants with CF and 82 healthy controls were enrolled (Table 1). The mean age for both groups was similar, but the percentage of males in the healthy controls was lower than that of the CF group (38% vs. 55%). More than 85% of participants with CF had a clinical diagnosis of pancreatic insufficiency, and 53% were receiving vitamin D supplementation compared with 22% of healthy controls. Approximately 50% and 40% of the participants with CF were homozygous and heterozygous for the cystic fibrosis transmembrane conductance regulator (CFTR) ΔF508 gene variation, respectively.
Table 1.
Characteristics of study participants
| Cross-Sectional Study | Pharmacokinetic Study | |||
|---|---|---|---|---|
| Participants with CF (n = 83) | Healthy Controls (n = 82) | Participants with CF (n = 5) | Healthy Controls (n = 5) | |
| Age (years) | 32.5 ± 11.2 | 24.8 ± 3.3 | ||
| Male | 45 (55%) | 31 (38%) | 4 (80%) | 1 (20%) |
| BMI (kg/m2) | 23.0 ± 3.4 | 25.5 ± 4.9 | 25.0 ± 3.1 | 22.4 ± 3.0 |
| Race | ||||
| White | 80 (96%) | 79 (96%) | 5 (100%) | 5 (100%) |
| Other | 3 (4%) | 3 (4%) | 0 (0%) | 0 (0%) |
| Season of Sample Collection | ||||
| Winter | 23 (37%) | 37 (45%) | 2 (40%) | 0 (0%) |
| Spring | 14 (17%) | 31 (38%) | 1 (20%) | 4 (80%) |
| Summer | 11 (13%) | 10 (12%) | 2 (40%) | 1 (20%) |
| Fall | 35 (42%) | 4 (5%) | 0 (0%) | 0 (0%) |
| Pancreatic Insufficient | 71 (86%) | --a | 3 (60%) | -- |
| Vitamin D Supplementation | 44 (53%) | 18 (22%) | -- | -- |
| FGF (pg/mL) | 44.4 ± 18.3 | 41.4 ± 11.8 | -- | -- |
| PTH (pg/mL) | 43.1 ± 19.0 | 40.6 ± 15.4 | -- | -- |
| CFTR Genotype | ||||
| Homozygous ΔF508 | 33 (40%) | -- | 1 (20%) | -- |
| Heterozygous ΔF508 | 42 (51%) | -- | 3 (60%) | -- |
| Other | 8 (10%) | -- | 1 (20%) | -- |
Results are reported as mean ± SD or count (%)
-- Not determined
3.2. Vitamin D Metabolite Concentrations in the Cross-Sectional Study
Only 35% of participants with CF had total 25(OH)D concentrations ≥ 30 ng/mL. Sixty-seven percent and 78% of participants with CF and healthy controls had total 25(OH)D concentrations ≥ 20 ng/mL, respectively. Mean ± standard deviation concentrations of 25(OH)D were comparable between participants with CF and healthy controls (26.7 ± 12.3 ng/mL and 27.7 ± 9.9 ng/mL, respectively; Table 2). Total 1α,25(OH)2D was systematically lower in participants with CF than in healthy controls, which persisted after adjustment for age, sex, season and total 25(OH)D (mean difference: −7.1 pg/mL; 95% CI: −11.0, −3.2; p < 0.001). Similar results were observed for 4β,25(OH)2D3 and 25(OH)D3-S. Concentrations of 24,25(OH)2D3 and of 25(OH)D3-G did not differ between the two groups before or after adjusted analyses.
Table 2:
Cross-sectional concentrations of 25-hydroxyvitamin D, its metabolites, and vitamin D binding proteins in participants with cystic fibrosis and healthy controls
| Unadjusted Data | Adjusted for age, sex, and season | Adjusted for age, sex, season, and 25(OH)D-specific metabolite | ||||||
|---|---|---|---|---|---|---|---|---|
| Participants with CF (n = 83) | Healthy Controls (n = 82) | CF vs. HealthyDifference (95% CI) | p-value | CF vs. Healthy Adjusted Difference (95% CI) | p-value | CF vs. Healthy Adjusted Difference (95% CI) | p-value | |
| Total Vitamin D Metabolites | ||||||||
| 25(OH)D (ng/mL) | 26.7 ± 12.3 | 27.7 ± 9.9 | −1.0 (−4.4, 2.4) | 0.57 | 1.2 (−2.2, 4.6) | 0.48 | --a | -- |
| 1α,25(OH)2D (pg/mL) | 43.6 ± 12.7 | 50.7 ± 13.0 | −7.1 (−11.0, −3.2) | <0.001 | −7.8 (−11.9, −3.6) | <0.001 | −7.9 (−12.0, −3.7) | <0.001 |
| Vitamin D3 Metabolites | ||||||||
| 25(OH)D3 (ng/mL) | 25.3 ± 12.0 | 26.8 ± 10.1 | −1.5 (−4.8, 1.9) | 0.39 | 0.6 (−2.8, 4.0) | 0.73 | -- | -- |
| Unbound 25(OH)D3 (pg/mL)b | 17.9 ± 8.4 | 18.2 ± 6.8 | −0.3 (−2.7, 2.1) | 0.80 | -- | -- | -- | -- |
| % Unbound 25(OH)D3 | 0.072 ± 0.009 | 0.06 9 ± 0.00 1 | 0.0023 (0.0003, 0.0043) | 0.027 | -- | -- | -- | -- |
| 1α,25(OH)2D3 (pg/mL) | 42.3 ± 13.3 | 49.5 ± 12.8 | −7.2 (−11.1, −3.2) | <0.001 | −7.9 (−12.2, −3.6) | <0.001 | −8.0 (−12.2, −3.7) | <0.001 |
| Unbound 1α,25(OH)2D3 (pg/mL)b | 0.51 ± 0.15 | 0.57 ± 0.13 | −0.058 (0.100, 0.015) | 0.009 | -- | -- | -- | -- |
| % Unbound 1α,25(OH)2D3 | 0.50 ± 0.06 | 0.49 ± 0.07 | 0.018 (0.001, 0.037) | 0.070 | -- | -- | -- | -- |
| 24,25(OH)2D3 (ng/mL) | 1.7 ± 1.2 | 1.7 ± 1.0 | 0.0 (−0.4, 0.3) | 0.81 | 0.1 (−0.2, 0.5) | 0.51 | 0.1 (−0.1, 0.2) | 0.44 |
| 4β,25(OH)2D3 (pg/mL)c | 52.1 ± 38.9 | 79.8 ± 60.5 | −27.6 (−43.2, −12.0) | <0.001 | −25.1 (−40.8, −9.3) | 0.002 | −26.6 (−40.3, −13.0) | <0.001 |
| 25(OH)D3−S (ng/mL) | 17.7 ± 11.6 | 30.1 ± 12.3 | −12.4 (−16.0, −8.7) | <0.001 | −11.2 (−14.9, −7.5) | <0.001 | −11.7 (−14.4, −8.9) | <0.001 |
| 25(OH)D3−G (ng/mL) | 1.9 ± 1.2 | 2.2 ± 1.6 | −0.3 (−0.7, 0.1) | 0.13 | −0.2 (−0.6, 0.2) | 0.37 | −0.2 (−0.5, 0.0) | 0.09 |
| Vitamin D2 Metabolites | ||||||||
| 25(OH)D2 (ng/mL) | 1.3 ± 5.4 | 0.9 ± 1.9 | 0.4 (−0.8, 1.7) | 0.49 | 0.6 (−1.0, 2.2) | 0.44 | -- | -- |
| 1α,25(OH)2D2 (pg/mL) | 1.3 ± 4.2 | 1.2 ± 2.5 | 0.1 (−0.9, 1.1) | 0.87 | 0.1 (−1.2, 1.4) | 0.86 | −0.4 (−0.8, 0.1) | 0.10 |
| Vitamin D Binding Proteins | ||||||||
| VDBP (μg/mL) | 264 ± 41 | 274 ± 56 | −10 (–25, 5) | 0.19 | -- | -- | -- | -- |
| Albumin (g/dL) | 3.9 ± 0.4 | 4.3 ± 0.3 | −0.4 (−0.5, −0.3) | <0.001 | -- | -- | -- | -- |
Results are reported as mean ± SD for unadjusted data and difference (95% CI) for adjusted models
-- Not determined
Unbound concentrations of 25(OH)D3 and 1α,25(OH)2D3 were calculated using previously published equations (21, 22)
For the CF group, n = 82 due to insufficient sample volume for one study participant
Serum VDBP concentrations were similar between the two groups, while serum albumin concentrations were lower in participants with CF than in healthy controls (Table 2). Although unbound 25(OH)D3 concentrations were comparable between participants with CF and healthy controls, participants with CF had a higher percent of unbound 25(OH)D3 than healthy controls (mean difference: 0.0023; 95% CI: 0.0003, 0.0043). Unbound 1α,25(OH)2D3 concentrations were 10% lower in participants with CF than in healthy controls (0.508 ± 0.151 vs. 0.566 ± 0.126 pg/mL; p < 0.05), while the percent of unbound 1α,25(OH)2D3 was comparable between the two groups.
3.3. Pharmacokinetic Study Participants
A total of 5 participants with CF and 5 healthy controls participated in the pharmacokinetic study (Table 1). Participants with CF were older and had a higher percentage of males than the healthy control group.
3.4. Pharmacokinetics of d6-25-hydroxyvitamin D3
The d6-25(OH)D3 and d6-24,25(OH)2D3 concentration vs. time profiles and pharmacokinetic parameters are presented in Figure 1 and Table 3, respectively. The peak concentration (Cmax) of d6-25(OH)D3 was observed at the earliest timepoint (15 min) for all study participants, after which d6-25(OH)D3 declined over time. No differences were noted in the mean clearance (CL), steady-state volume of distribution (Vss), or terminal half-life (t1/2) of d6-25(OH)D3 between participants with CF and healthy controls (p > 0.05 for all). The coefficient of variation (CV%) for d6-25(OH)D3 pharmacokinetic parameters was less than 20% for both groups.
Figure 1.

Deuterated 25-hydroxyvitamin D3 and 24,25-dihydroxyvitamin D3 concentrations were similar between CF and healthy controls over time. d6-25(OH)D3 concentrations, dose-adjusted to 25 μg d6-25(OH)D3, are presented as circles and solid lines, while d6-24,25(OH)2D3 concentrations are presented as squares and dashed lines. Group means are presented for participants with CF (closed symbols) and healthy controls (open symbols). The vertical whiskers indicate the standard deviation in each direction.
Table 3:
Summary of pharmacokinetic (PK) parameters after intravenous administration of d6-25-hydroxyvitamin D3 in participants with cystic fibrosis and healthy controls
| Parametera | Participants with CF (n=5) | Healthy Controls (n=5) | p-value |
|---|---|---|---|
| Actual dose d6-25(OH)D3 (μg) | 26.4 ± 3.6 | 22.0 ± 3.2 | 0.11 |
| d6-25(OH)D3 Dose-Adjusted PK Parametersb | |||
| Cmax (ng/mL) | 8.8 ± 1.4 | 9.4 ± 1.9 | 0.84 |
| AUC0-last (ng·day/mL) | 58.3 ± 9.7 | 67.2 ± 8.3 | 0.22 |
| AUC0−∞ (ng·day/mL) | 65.1 ± 12.5 | 73.9 ± 9.0 | 0.31 |
| Extrapolated AUC (%) | 10.0 ± 3.6 | 8.9 ± 1.6 | 0.69 |
| CL (mL/day) | 397 ± 73 | 342 ± 41 | 0.31 |
| VSS (L) | 8.4 ± 1.4 | 7.2 ± 1.1 | 0.10 |
| t1/2 (day) | 16.2 ± 3.3 | 15.8 ± 1.6 | 0.84 |
| d6-24,25(OH)2D3 PK Parameters | |||
| Cmax (ng/mL) | 0.16 ± 0.02 | 0.13 ± 0.02 | 0.07 |
| AUC0-last (ng·day/mL) | 5.9 ± 1.2 | 4.8 ± 0.9 | 0.15 |
Results are reported as mean ± SD
Pharmacokinetic parameters for d6-25(OH)D3 were calculated using dose-normalized d6-25(OH)D3 serum concentrations adjusted to 25 μg d6-25(OH)D3.
The mean Cmax of d6-24,25(OH)2D3 was comparable between participants with CF and healthy controls and peaked at a median (interquartile range) of 14 (4–14) days for both groups. The d6-24,25(OH)2D3 AUC0-last and molar AUC ratio did not differ between participants with CF and healthy controls. Due to variability in the d6-24,25(OH)2D3 data, the terminal half-life and AUC0−∞ could not be estimated.
4. Discussion
The present investigation is one of the first to evaluate a comprehensive set of vitamin D metabolites and the pharmacokinetics of 25(OH)D in adults with CF. Only 35% of participants with CF had total 25(OH)D concentrations greater than or equal to the guideline-recommended threshold of 30 ng/mL despite over 50% of them taking vitamin D supplements. Of the metabolites studied, concentrations of total 1α,25(OH)2D, 4β,25(OH)2D3, and 25(OH)D3-S were lower in participants with CF than in healthy controls, even after covariate adjustment. There were no differences in the pharmacokinetics of d6-25(OH)D3 or in the formation of d6-24,25(OH)2D3 between the two groups.
25(OH)D is the most studied vitamin D metabolite in CF, and our results are consistent with that of others reporting low serum concentrations despite routine vitamin D supplementation (2, 4–7). The downstream metabolites of 25(OH)D, however, have received significantly less attention. Greer et al. reported that total 1α,25(OH)2D concentrations were lower in children, adolescents, and adults with CF than in healthy controls despite comparable 25(OH)D concentrations, similar to our findings in adults (24). Weisman et al. reported no differences in 24,25(OH)2D3 concentrations between children with and without CF (2.2 ng/mL ± 0.6 and 2.6 ± 1.3 ng/mL, respectively), but observed concentrations higher than seen in our study, possibly due to differences in analytical technique or study population (25). To our knowledge, this is the first study to assess 24,25(OH)2D3, 4β,25(OH)2D3, 25(OH)D3-G and 25(OH)D3-S concentrations in adults with CF.
In a mechanism thought to protect against vitamin D toxicity, 25(OH)D3 is metabolized by CYP24A1 into 24,25(OH)2D3, the predominant downstream metabolite of 25(OH)D3 in the serum (11, 18). We hypothesized that increased 25(OH)D3 clearance and 24,25(OH)2D3 formation could contribute to low 25(OH)D concentrations in CF, but did not observe significant differences in these pharmacokinetic parameters between participants with CF and healthy controls. While our study was likely underpowered to detect more modest differences in pharmacokinetic parameters between the two groups, it seems reasonable to conclude that altered 25(OH)D3 clearance does not contribute substantially to low 25(OH)D concentrations in persons with CF.
The lower concentrations of 25(OH)D3-S in participants with CF than in healthy controls offer some insight into potential mechanisms for low 25(OH)D in CF. In the liver, 25(OH)D3 undergoes sulfation by sulfotransferase 2A1 (SULT2A1) into 25(OH)D3-S, which has been hypothesized to act as a vitamin D reservoir as it is protected from further hydroxylation into 24,25(OH)2D3 or 1,25(OH)2D3 unlike 25(OH)D3 (26). 25(OH)D3-S may be excreted into bile, reabsorbed in the small intestine, and deconjugated back into 25(OH)D3 (14, 27). Excess biliary excretion or impaired enterohepatic recirculation of 25(OH)D3-S may therefore result in low concentrations of 25(OH)D3-S and 25(OH)D3 in CF and requires further investigation.
It is less clear how our findings of low 1α,25(OH)2D and 4β,25(OH)2D3 in participants with CF explain low 25(OH)D in CF, as they suggest reduced formation of these metabolites from 25(OH)D. Nonetheless, these observations may offer some insight on the pathophysiology of their respective pathways in CF. As the biologically active form of vitamin D, 1α,25(OH)2D concentrations are tightly regulated in healthy persons, in particular by fibroblast growth factor-23 (FGF-23) and parathyroid hormone (PTH). Given that concentrations of FGF-23 and PTH were similar between participants with CF and healthy controls, other factors may be responsible for reduced 1α-hydroxylase activity in CF, including defects to the CFTR, which is abundantly expressed in the kidney (28). Similarly, patients with CF may experience down-regulation of hepatic CYP3A4, the enzyme responsible for the formation of 4β,25(OH)2D3 (13). Whether these changes are related to the pathology of CF, interactions with medications (ivacaftor, for example, is also metabolized by CYP3A4 (29)), or other mechanisms remain to be explored.
Strengths of this study include the comprehensive evaluation of vitamin D metabolites using precise and accurate measurements, and the first-ever use of gold-standard pharmacokinetic methods to determine 25(OH)D3 clearance in CF. There are a few limitations. First, due to the cross-sectional nature of the study and the lack of clinical outcomes, we could not determine whether the measured differences in metabolites of 25(OH)D have clinical implications. Second, we did not directly measure enzymatic activity. It is possible that low concentrations of 1α,25(OH)2D, 4β,25(OH)2D3, and 25(OH)D3-S are not due to reduced formation, but rather increased elimination. Third, we did not measure 3-epi-25-hydroxyvitamin D3, an increasingly recognized epimer of 25(OH)D3, which circulates at approximately 4–5% of 25(OH)D3 concentrations (30, 31). As we found measured 25(OH)D3 clearance to be similar between CF and controls, potential differences in C3 epimerization of 25(OH)D3 seem insufficient to explain low 25(OH)D in CF, though the importance of C3 epimers of vitamin D in CF remains to be determined. Fourth, we did not measure vitamin D3, the precursor to 25(OH)D3, and cannot make any inferences with regards to this metabolic pathway.
5. Conclusions
Compared with healthy participants, those with CF had lower total 1α,25(OH)2D, 4β,25(OH)2D3 and 25(OH)D3-S, perhaps as a consequence of decreased formation in CF. As there were no differences in the systemic clearance or half-life of d6-25(OH)D3 between our two study groups, increased 25(OH)D3 elimination or formation of 24,25(OH)2D3 are unlikely to contribute to low 25(OH)D concentrations in persons with CF. Other factors that may explain 25(OH)D deficiency in CF, such as altered absorption of vitamin D, decreased hepatic 25-hydroxylation, or losses through decreased enterohepatic recirculation need further investigation. Identifying the precise mechanisms involved may improve how we assess and treat vitamin D-related complications of CF.
Supplementary Material
HIGHLIGHTS.
Assessed 8 vitamin D metabolites in participants with cystic fibrosis and controls
Total 25(OH)D was comparable between participants with cystic fibrosis and controls
Total 1α,25(OH)2D, 4β,25(OH)2D3, 25(OH)D3-3-sulfate were lower in cystic fibrosis
Clearance of 25(OH)D3 did not differ between groups in a pharmacokinetic study
Acknowledgements
We thank the participants of the CLEAR-CF and SKS studies for their participation.
Funding:
This work was supported in part by grants from the National Institutes of Health: P30 DK089507, P30 DK035816, P30 DK017047, R01 DK099199, T32 GM007750 (to MCB), T32 DK007467-36 (to SH), and by an unrestricted fund from the Northwest Kidney Centers.
Abbreviations:
- 1α,25(OH)2D
1α,25-dihydroxyvitamin D
- 1α,25(OH)2D2
1α,25-dihydroxyvitamin2
- 1α,25(OH)2D3
1α,25-dihydroxyvitamin D3
- 24,25(OH)2D3
24,25-dihydroxyvitamin D3
- 25(OH)D
25-hydroxyvitamin D
- 25(OH)D2
25-hydroxyvitamin D2
- 25(OH)D3
25-hydroxyvitamin D3
- 25(OH)D3-G
25-hydroxyvitamin D3-3-glucuronide
- 25(OH)D3-S
25-hydroxyvitamin D3-3sulfate
- 4β,25(OH)2D3
4β,25-dihydroxyvitamin D3
- AUC0−∞
area under the concentration-time curve
- AUC0-last
area under the concentration-time curve to the last observed time point
- CFTR
cystic fibrosis transmembrane conductance regulator gene
- CL
systemic clearance
- Cmax
maximum observed concentration
- CF
cystic fibrosis
- d6-24,25(OH)2D3
deuterium-labeled 24,25-dihydroxyvitamin D3
- d6-25(OH)D3
deuterium-labeled 25-hydroxyvitamin D3
- FGF-23
fibroblast growth factor-23
- PK
pharmacokinetic
- PTH
parathyroid hormone
- SULT2A1
sulfotransferase 2A1
- t1/2
elimination half-life
- Tmax
time of maximum concentration
- VDBP
vitamin D binding protein
- Vss
steady-state volume of distribution
Footnotes
Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.
Declarations of interest: none
References
- 1.Daley T, Hughan K, Rayas M, Kelly A, Tangpricha V. Vitamin D deficiency and its treatment in cystic fibrosis. Journal of Cystic Fibrosis. 2019;18:S66–S73. [DOI] [PubMed] [Google Scholar]
- 2.Donovan DS, Jr., Papadopoulos A, Staron RB, Addesso V, Schulman L, McGregor C, et al. Bone mass and vitamin D deficiency in adults with advanced cystic fibrosis lung disease. Am J Respir Crit Care Med. 1998;157(6 Pt 1):1892–9. [DOI] [PubMed] [Google Scholar]
- 3.Putman MS, Anabtawi A, Le T, Tangpricha V, Sermet-Gaudelus I. Cystic fibrosis bone disease treatment: Current knowledge and future directions. J Cyst Fibros. 2019;18 Suppl 2:S56–S65. [DOI] [PubMed] [Google Scholar]
- 4.Tangpricha V, Kelly A, Stephenson A, Maguiness K, Enders J, Robinson KA, et al. An update on the screening, diagnosis, management, and treatment of vitamin D deficiency in individuals with cystic fibrosis: evidence-based recommendations from the Cystic Fibrosis Foundation. J Clin Endocrinol Metab. 2012;97(4):1082–93. [DOI] [PubMed] [Google Scholar]
- 5.Li L, Somerset S. Dietary intake and nutritional status of micronutrients in adults with cystic fibrosis in relation to current recommendations. Clin Nutr. 2016;35(4):775–82. [DOI] [PubMed] [Google Scholar]
- 6.Chavasse RJ, Francis J, Balfour-Lynn I, Rosenthal M, Bush A. Serum vitamin D levels in children with cystic fibrosis. Pediatr Pulmonol. 2004;38(2):119–22. [DOI] [PubMed] [Google Scholar]
- 7.Rovner AJ, Stallings VA, Schall JI, Leonard MB, Zemel BS. Vitamin D insufficiency in children, adolescents, and young adults with cystic fibrosis despite routine oral supplementation. Am J Clin Nutr. 2007;86(6):1694–9. [DOI] [PubMed] [Google Scholar]
- 8.Bikle DD. Vitamin D metabolism, mechanism of action, and clinical applications. Chem Biol. 2014;21(3):319–29. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Hsu S, Zelnick LR, Lin YS, Best CM, Kestenbaum B, Thummel KE, et al. Differences in 25-Hydroxyvitamin D Clearance by eGFR and Race: A Pharmacokinetic Study. J Am Soc Nephrol. 2021;32(1):188–98. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Cheng JB, Motola DL, Mangelsdorf DJ, Russell DW. De-orphanization of cytochrome P450 2R1: a microsomal vitamin D 25-hydroxilase. The Journal of biological chemistry. 2003;278(39):38084–93. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Tuckey RC, Cheng CYS, Slominski AT. The serum vitamin D metabolome: What we know and what is still to discover. J Steroid Biochem Mol Biol. 2019;186:4–21. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Pauwels S, Jans I, Billen J, Heijboer A, Verstuyf A, Carmeliet G, et al. 1β,25-Dihydroxyvitamin D(3): A new vitamin D metabolite in human serum. J Steroid Biochem Mol Biol. 2017;173:341–8. [DOI] [PubMed] [Google Scholar]
- 13.Wang Z, Lin YS, Zheng XE, Senn T, Hashizume T, Scian M, et al. An inducible cytochrome P450 3A4-dependent vitamin D catabolic pathway. Mol Pharmacol. 2012;81(4):498–509. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Wong T, Wang Z, Chapron BD, Suzuki M, Claw KG, Gao C, et al. Polymorphic Human Sulfotransferase 2A1 Mediates the Formation of 25-Hydroxyvitamin D3–3-O-Sulfate, a Major Circulating Vitamin D Metabolite in Humans. Drug Metab Dispos. 2018;46(4):367–79. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Wang Z, Wong T, Hashizume T, Dickmann LZ, Scian M, Koszewski NJ, et al. Human UGT1A4 and UGT1A3 conjugate 25-hydroxyvitamin D3: metabolite structure, kinetics, inducibility, and interindividual variability. Endocrinology. 2014;155(6):2052–63. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Mailhot G Vitamin D bioavailability in cystic fibrosis: a cause for concern? Nutrition Reviews. 2012;70(5):280–93. [DOI] [PubMed] [Google Scholar]
- 17.Moustaki M, Loukou I, Priftis KN, Douros K. Role of vitamin D in cystic fibrosis and non-cystic fibrosis bronchiectasis. World J Clin Pediatr. 2017;6(3):132–42. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Bosworth CR, Levin G, Robinson-Cohen C, Hoofnagle AN, Ruzinski J, Young B, et al. The serum 24,25-dihydroxyvitamin D concentration, a marker of vitamin D catabolism, is reduced in chronic kidney disease. Kidney Int. 2012;82(6):693–700. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Jing J, Isoherranen N, Robinson-Cohen C, Petrie I, Kestenbaum BR, Yeung CK. Chronic Kidney Disease Alters Vitamin A Homeostasis via Effects on Hepatic RBP4 Protein Expression and Metabolic Enzymes. Clin Transl Sci. 2016;9(4):207–15. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Nadler SB, Hidalgo JH, Bloch T. Prediction of blood volume in normal human adults. Surgery. 1962;51(2):224–32. [PubMed] [Google Scholar]
- 21.Bikle DD, Gee E, Halloran B, Kowalski MA, Ryzen E, Haddad JG. Assessment of the free fraction of 25-hydroxyvitamin D in serum and its regulation by albumin and the vitamin D-binding protein. J Clin Endocrinol Metab. 1986;63(4):954–9. [DOI] [PubMed] [Google Scholar]
- 22.Schwartz JB, Lai J, Lizaola B, Kane L, Weyland P, Terrault NA, et al. Variability in free 25(OH) vitamin D levels in clinical populations. J Steroid Biochem Mol Biol. 2014;144 Pt A:156–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Bikle DD, Malmstroem S, Schwartz J. Current Controversies: Are Free Vitamin Metabolite Levels a More Accurate Assessment of Vitamin D Status than Total Levels? Endocrinol Metab Clin North Am. 2017;46(4):901–18. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Greer RM, Buntain HM, Potter JM, Wainwright CE, Wong JC, O’Rourke PK, et al. Abnormalities of the PTH-vitamin D axis and bone turnover markers in children, adolescents and adults with cystic fibrosis: comparison with healthy controls. Osteoporos Int. 2003;14(5):404–11. [DOI] [PubMed] [Google Scholar]
- 25.Weisman Y, Reiter E, Stern RC, Root A. Serum concentrations of 25-hydroxyvitamin D and 24,25-dihydroxyvitamin D in patients with cystic fibrosis. J Pediatr. 1979;95(3):416–8. [DOI] [PubMed] [Google Scholar]
- 26.Tuckey RC, Cheng CYS, Li L, Jiang Y. Analysis of the ability of vitamin D3-metabolizing cytochromes P450 to act on vitamin D3 sulfate and 25-hydroxyvitamin D3 3-sulfate. The Journal of Steroid Biochemistry and Molecular Biology. 2023;227:106229. [DOI] [PubMed] [Google Scholar]
- 27.Gao C, Liao MZ, Han LW, Thummel KE, Mao Q. Hepatic Transport of 25-Hydroxyvitamin D(3) Conjugates: A Mechanism of 25-Hydroxyvitamin D(3) Delivery to the Intestinal Tract. Drug Metab Dispos. 2018;46(5):581–91. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Souza-Menezes J, da Silva Feltran G, Morales MM. CFTR and TNR-CFTR expression and function in the kidney. Biophys Rev. 2014;6(2):227–36. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.van der Meer R, Wilms EB, Sturm R, Heijerman HGM. Pharmacokinetic interactions between ivacaftor and cytochrome P450 3A4 inhibitors in people with cystic fibrosis and healthy controls. J Cyst Fibros. 2021;20(5):e72–e6. [DOI] [PubMed] [Google Scholar]
- 30.Cashman KD, Kinsella M, Walton J, Flynn A, Hayes A, Lucey AJ, et al. The 3 epimer of 25-hydroxycholecalciferol is present in the circulation of the majority of adults in a nationally representative sample and has endogenous origins. J Nutr. 2014;144(7):1050–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Al-Zohily B, Al-Menhali A, Gariballa S, Haq A, Shah I. Epimers of Vitamin D: A Review. Int J Mol Sci. 2020;21(2). [DOI] [PMC free article] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.
